WO1998052553A1 - Procedes de traitement d'etats associes au lactosylceramide - Google Patents

Procedes de traitement d'etats associes au lactosylceramide Download PDF

Info

Publication number
WO1998052553A1
WO1998052553A1 PCT/US1998/009958 US9809958W WO9852553A1 WO 1998052553 A1 WO1998052553 A1 WO 1998052553A1 US 9809958 W US9809958 W US 9809958W WO 9852553 A1 WO9852553 A1 WO 9852553A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
galt
laccer
inhibitor compound
lactosylceramide
Prior art date
Application number
PCT/US1998/009958
Other languages
English (en)
Inventor
Subroto Chatterjee
Original Assignee
Johns Hopkins University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Johns Hopkins University filed Critical Johns Hopkins University
Priority to CA2290742A priority Critical patent/CA2290742C/fr
Priority to AU76866/98A priority patent/AU742468B2/en
Priority to JP55046098A priority patent/JP2001526683A/ja
Priority to EP98924776A priority patent/EP0996433A4/fr
Publication of WO1998052553A1 publication Critical patent/WO1998052553A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the present invention includes methods for treating conditions modulated by lactosylceramide and, more particularly, to the use of one or more compounds that inhibit UDP-galactose, GlcCer, ⁇ l->4 galactosyltransferase (GalT-2) to treat a subject suffering from or susceptible to a condition caused or contributed to by lactosylceramide.
  • the present invention also relates to methods for detecting and analyzing compounds with therapeutic capacity to treat such conditions.
  • Uncontrolled cell proliferation relates to conditions impacting the heart, kidney, liver and other organs.
  • uncontrolled cell proliferation is indicative of diseases such as certain vasculopathies, e.g., atherosclerosis, and pathologies involving neovascularization, tumor or cyst development, e.g., polycystic kidney disease and post-surgical keloid formation.
  • diseases such as certain vasculopathies, e.g., atherosclerosis, and pathologies involving neovascularization, tumor or cyst development, e.g., polycystic kidney disease and post-surgical keloid formation.
  • uncontrolled proliferation of vascular cells can significantly contribute to disease by occluding blood flow and enhancing vessel remodeling.
  • Certain post-surgical disorders such as restenosis are particularly affected by uncontrolled vascular cell proliferation. See generally Harrison 's Principles of Internal Medicine, (1991) 12 th ed. McGraw-Hill, Inc.; and Cole, B.R. (1990) in The Cystic Kidney
  • Invasive surgical procedures have been developed to alleviate certain diseases and post-surgical disorders.
  • certain surgical techniques involving angioplasty, and particularly balloon angioplasty have been developed to enhance vascular flow.
  • angioplasty is often accompanied by side effects such as restenosis.
  • restenosis is recognized as a serious post-surgical complication of angioplasty. See Landau, C, et al. (1994) N. Eng. J. Med., 330:981 and references cited therein.
  • probucol is a recognized therapeutic agent that has been reported to reduce restenosis in some patients.
  • existing probucol- based therapies are believed to be ineffective due to unsatisfactory patient tolerance and insufficient reduction of stenoses. See e.g., Tardif, J. C. et al., (1997) N. Eng. J. Med, 337:365-372; Ferns, GAA. et al. (1992) PNAS (USA) 89:11312 and references cited therein.
  • GSLs glycosphingolipids
  • the GSLs are believed to impact lipid storage diseases, particularly glycosphingolipidoses and perhaps other lipid-related pathologies. See e.g., Chatterjee, S., Biochem. Biophys. Res Comm. (1991) 181:554; Hakomori, S.I. (1983) in Sphingolipid Chemistry, eds. Kanfer, J. ⁇ . and Hakomori, S.I. (Plenum Press, New York) and references cited therein. Certain biochemical steps relating to GlcCer and LacCer have been disclosed.
  • one step involves synthesis of GlcCer by coupling UDP-glucose to ceramide in a reaction catalyzed by UDP-glucose glucosyltransferase (GlcT-1).
  • Another step converts the GlcCer to LacCer using UDP-galactose, GlcCer, ⁇ l->4 galactosyltransferase (GalT-2). See e.g., Chatterjee et al. supra. Attempts have been made to inhibit biochemical steps involving GlcT-1.
  • D-PDMP D-l-phenyl-2-decanolylamino-3-morpholino-l- propanol
  • D-PDMP D-l-phenyl-2-decanolylamino-3-morpholino-l- propanol
  • the mechanism of PDMP has been reported to be unclear. See e.g., Felding-Habermann, B., et al. (1991) Biochemistry 29:6314; Shukla, G.S. et al. Biochem. Biophys. Acta. (1991) 1083:101; Inokuchi, J. et al., J. Lipid. Res. (1987) 28:565; Chatterjee, S., supra.
  • Specified morpholinoceramides also have been disclosed as GlcT-1 inhibitors. See Carson, K. and B. Ganem (1994) Tetrahedron Lets. 35:2659.
  • LacCer Other cell functions are believed to play a role in conditions modulated by LacCer. For example, uncontrolled cell adhesion is believed to effect specified immune responses such as allergic reactions and host rejection of foreign tissue.
  • IMM-1 intercellular adhesion molecule- 1
  • VCAM-1 vascular cell adhesion molecule-1
  • Kume Kume, N. et al. (1992) J. Clin. Invest. 90:1138; Iademarco, M.F. et al. (1995) J. Clin. Invest. 95:264; Carlos, T. et al. Blood (1991) 77:2266; Nagel, T. et al. (1994) J. Clin. Invest. 94:885; and Cybulsky, M.I. and Gimbrone MA. (1991) Science 251:788.
  • GSLs also have been reported to be cell receptors for bacterial toxins. For example, cellular uptake of cholera toxin is believed to be enhanced by a membrane-associated GSLs.
  • the invention provides methods for treatment of proliferative disorders such as vasculopathies, e.g.
  • Atherosclerosis and restenosis pathologies involving neovascularization; tumor or cyst development, e.g., polycystic kidney disease and post-surgical keloid formation; inflammatory diseases involving a proinflammatory cytokine such as TNF- ⁇ or interleukin-6; and lipid storage diseases such as Gaucher's disease.
  • a proinflammatory cytokine such as TNF- ⁇ or interleukin-6
  • lipid storage diseases such as Gaucher's disease.
  • Therapies of the invention are particularly effective for the treatment and prevention of undesired vascular restenoses.
  • a near absence of intimal proliferation was observed at the site of balloon angioplasty in the test subject (rabbit), whereas a control subject exhibited significant intimal proliferation. See the results set forth in the examples which follow.
  • LacCer-modulated diseases that can be treated in accordance with the invention also include lipid storage diseases (i.e. glycospingolipodoses) such as Gaucher's disease, cholesterol storage disease and the like.
  • lipid storage diseases i.e. glycospingolipodoses
  • Gaucher's disease i.e. Gaucher's disease, cholesterol storage disease and the like.
  • Additional disorders that can be treated in accordance with the invention include bacterial infections, particularly those infections involving production of a toxin, such as an exotoxin that can specifically bind LacCer, e.g. Neisseria gonorrhoeae. See Paruchuri, D.K. et al. (1990) Proc. Natl. Acad. Sci. (USA), 87:333- 337.
  • bacterial infections particularly those infections involving production of a toxin, such as an exotoxin that can specifically bind LacCer, e.g. Neisseria gonorrhoeae. See Paruchuri, D.K. et al. (1990) Proc. Natl. Acad. Sci. (USA), 87:333- 337.
  • Therapeutic methods of the invention in general comprise administering to a subject, particularly a mammal such as a primate, especially a human, a therapeutically effective amount of a compound that can inhibit GalT-2 activity.
  • a subject particularly a mammal such as a primate, especially a human
  • a therapeutically effective amount of a compound that can inhibit GalT-2 activity Preferably, an administered compound inhibits cell proliferation by at least about 15% or 25% in a standard in vitro cell proliferation assay. Examples of such an assay are described below.
  • the administered compound exhibits an IC 50 of at least about 500 ⁇ M in a standard in vitro GalT-2 assay as defined below, more preferably an IC 50 of about 100 ⁇ M or less, still more preferably an IC50 of about 1-10 ⁇ M or less in a standard in vitro GalT-2 assay as defined below.
  • Such compounds that can inhibit GalT-2 activity are generally referred to herein as "GalT-2 inhibitor compounds" or other similar term.
  • R, R , R and R are as defined below; and pharmaceutically acceptable salts of such compounds.
  • inhibitor compounds for use in the therapeutic methods of the invention include: 1 -phenyl-2-decanoylamino-3-mo holino- 1 -propanol; 1 -phenyl-2-hexadecanoylamino-3-morpholino- 1 -propanol; 1 -phenyl-2-hexadecanoylamino-3-piperidino- 1 -propanol; 1 -phenyl-2-hexadecanoylamino-3-pyrrolidino- 1 -propanol; 1 -morpholino-2-hexadecanoylamino-3-hydroxyoctadec-4,5-ene; and 1 -pyrrolidino-2-hexadecanoylamino-3-hydroxyoctadec-4,5-ene.
  • Especially preferred inhibitor compounds for use in the methods of the invention are (lR,2R)-l-phenyl-2-decanoylamino-3-morpholino-l-propanol (D- PDMP) and tra «s-(2R,3R)- 1 -pyr ⁇ olidino-2-hexadecanoylamino-3-hydroxyoctadec- 4,5-ene.
  • Other suitable GalT-2 inhibitor compounds can be readily identified by simple testing, e.g. by in vitro testing of a candidate inhibitor compound relative to a control for the ability to inhibit GalT-2 activity, e.g. by at least 10% more than a control.
  • the invention further relates to methods of detecting and analyzing compounds that inhibit GalT-2 and exhibit therapeutic capacity to treat or prevent the above-described conditions.
  • Preferred detection and analysis methods include both in vitro and in vivo assays to determine the therapeutic capacity of agents to modulate LacCer-responsive cells.
  • Preferred in vitro detection assays involve one or more steps associated with LacCer-related pathways.
  • Such assays include the following steps 1) through 4):
  • the in vitro assays of the present invention can be conducted with nearly any population of cells responsive to LacCer including a lysate of such cells or tissue, or a substantially purified fraction of the lysate.
  • Suitable LacCer responsive cells that may be employed in the assay include, e.g., cells associated with vascular intima, particularly primary and/or immortalized endothelial and smooth muscle cells, as well as certain immune cells such as leukocytes.
  • Preferred LacCer lysates or subcellular fractions include GalT-2.
  • the in vitro detection assays of the invention can be adapted in accordance with intended use. For example, as noted above, it has been found that LacCer manifests changes in certain cell functions such as cell proliferation and adhesion.
  • the standard in vitro assay above can be modified at step 3) to include measuring cell proliferation or adhesion in response to the added LacCer, and to determine any effect of the GalT-2 inhibitor compound on the cell function.
  • the known or candidate GalT-2 inhibitor compound tested in the assays can be employed as a sole active agent or in combination with other agents including other GalT-2 inhibitor compounds to be tested.
  • the in vitro assays are performed with a suitable control assay usually comprising the same test conditions as in the steps above, but without adding the GalT-2 inhibitor compound to the medium.
  • a candidate GalT-2 inhibitor compound can be identified as exhibiting desired activity by exhibiting at least about 10 percent greater activity relative to the control; more preferably at least about 20% greater activity relative to the control assay; and still more preferably at least about 30%, 40%, 50%, 60%, 70, 80%, 100%, 150% or 200% greater activity relative to the control.
  • the invention also provides assays to detect a LacCer-responsive cell which cells may be used, e.g., in an assay of the invention as described above. For example, a potentially LacCer-responsive cell can be contacted by LacCer and then a desired cell molecule or function in a LacCer-related protein as discussed previously is measured as a function of the amount of LacCer added.
  • the cell is deemed responsive to LacCer if the assay employed shows at least about 10%, preferably at least about 20%, more preferably at least about 50%, and still more preferably at least about 75% or 100% change in the activity (relative to a control) of the molecule or cell function as determined by the assays provided herein.
  • the assays can be used to identify LacCer-responsiveness in a variety of cells or tissues, including cultured cells (i.e., primary cells or immortalized cell lines) and organs.
  • the invention also provides in vivo assays to determine the therapeutic capacity of a known or candidate GalT-2 inhibitor compound to modulate cell functions impacted by LacCer, e.g. cell proliferation and adhesion.
  • the monitored cell function suitably may be pre-existing in the test animal, or the cell function may be induced, e.g., by an invasive surgical procedure such as angioplasty.
  • Cell functions that can be suitably assayed in these methods include, e.g., vascular cell proliferation and adhesion as well as vessel remodeling.
  • the in vivo assays of the present invention can be modified in a number of ways as needed.
  • the vessel subjected to analysis is assayed in vitro following removal from the animal or assayed in vivo if desired.
  • the GalT-2 inhibitor compound is administered to the animal either as a sole active agent or in combination with other active compounds (e.g., probucol), including other GalT-2 inhibitor compounds to be tested.
  • activity of the GalT-2 inhibitor compound in a given in vivo assay is compared to a suitable control (e.g., a sham-operated animal) in which the assay is conducted the same as the test assay but without administering the GalT-2 inhibitor compound to the test subject.
  • a suitable control e.g., a sham-operated animal
  • test subjects can be employed, particularly mammals such as rabbits, primates, various rodents and the like.
  • the detection assays can be conducted in a wide variety of LacCer-responsive cells, tissues and organs. Further, the assays can detect useful GalT-2 inhibitor compounds by measuring the activity of target molecules and functions in LacCer-related pathways.
  • the present assays can measure activity in several cell, tissue and organ settings.
  • use of multiple detection assays e.g., a combination of the in vitro and/or in vivo assays
  • a single GalT-2 inhibitor compound can extend the selectivity and sensitivity of detection as desired.
  • in vitro assays of the invention can efficiently perform multiple analyses, thereby enhancing efficiency and probability of identifying GalT-2 inhibitor compounds with therapeutic capacity. This is especially useful when large numbers of compounds need to be tested.
  • libraries of GalT-2 inhibitor compounds can be made by standard synthetic methods including combinatorial-type chemistry manipulations and then tested in accord with the invention.
  • LacCer-related steps are "downstream" of GalT-2, and therefore the assays include molecules and cell functions that are active downstream of GalT-2. Accordingly, modest but significant changes in GalT-2 activity can be registered as readily testable signals. Other aspects of the invention are discussed below.
  • Figs. 1 A-C are graphs illustrating effects of D-PDMP on tumor necrosis factor- ⁇ (TNF- ⁇ ) induced ICAM-1 expression and GalT-2 activity in endothelial cells.
  • Figs. 2 A-C are photomicrographs of hematoxylin eosin-stained rabbit aorta sections following balloon angioplasty and treatment with D-PDMP.
  • Figs. 3A-C are graphs showing that LacCer stimulates superoxide generation in Eahy 926 cells (a human endothelial cell line).
  • Figs. 4A-B are graphs showing that LacCer stimulates activity of NADPH oxidase in Eahy 926 cells (a human endothelial cell line). Similar results have been obtained with a human umbilical cell line.
  • Fig. 5A-B are representations of gel mobility assays illustrating effect of LacCer on the expression of NF-kB in Eahy 926 cells.
  • Figs. 6 A-B are graphs showing effects of various glycosphingolipids on ICAM-1 expression in Eahy 926 cells.
  • Figs. 6C-D are photographs showing effects of various glycosphingolipids on ICAM-1 expression in Eahy 926 cells.
  • Figs. 7A-B are graphs depicting effects of DPI, NAC and superoxide dismutase on LacCer-induced superoxide generation and ICAM-1 expression.
  • Figs. 8A-C are graphs showing adenovirus-mediated overexpression of superoxide dismutase and ICAM-1 expression by LacCer in Eahy 926 cells.
  • Figs. 9A-D are drawings of color photographs (color indicated by shading) showing that LacCer stimulates neutrophil adhesion to Eahy 926 cells and NAC and DPI abrogate that phenomenon.
  • Figs. 10A-L are graphs showing LacCer upregulation of Mac- 1 expression in human PMNs.
  • Figs. 11 A-B are graphs illustrating PMN ROM generation under various incubation conditions.
  • Figs. 12 A-B are graphs showing that phospholipase A 2 inhibition reduced LacCer upregulation of Mac- 1 on PMN.
  • Fig. 12C is a graph showing inhibition of LacCer-induced superoxide generation by various inhibitors.
  • Fig. 13 is a graph showing that LacCer increased PMN adhesion to unstimulated endothelial cells via a CDl lb/CD18, Mac- 1 -dependent mechanism.
  • Fig. 14 is a graph showing that LacCer increased PMN adhesion to endothelial cells via a CDl l/CD18-dependent mechanism as a function of incubation time with LacCer.
  • Fig. 15 is a graph depicting quinacrine and bromophenacyl bromide (BPB)- reduced PMN adhesion to endothelial cells.
  • Fig. 16 is a graph showing that LacCer increased [ H] arachidoic acid release from labeled PMNs.
  • Fig. 17 is a model depicting LacCer-mediated redox signaling leading to ICAM-1 expression in endothelial cells and adhesion to neutrophils.
  • Fig. 18 is a model depicting utilization of Ox-LDL, LacCer, and lipid second messenger in the proliferation of H-ASMC.
  • Fig. 19 is a model depicting the role of LacCer as a lipid second messenger and its role in atherosclerosis and use of D-PDMP to abrogate this phenomenon.
  • the present invention features therapeutic methods for treatment and prevention of conditions modulated by LacCer.
  • the treatment methods of the invention generally include administering a therapeutically effective amount of a GalT-2 inhibitor compound to a subject, preferably a patient in need of such treatment.
  • LacCer is a cell signaling molecule that can modulate various diseases, post-surgical disorders and bacterial infections. That is, changes in cell levels of LacCer alter the development or severity of those diseases, post-surgical disorders and bacterial infections. More particularly, it has been unexpectedly found that in LacCer-responsive cells, LacCer functions as a signal molecule to effect changes in certain cell steps (sometimes referred to herein as "LacCer-related steps” or “LacCer-related pathways”). LacCer-related pathways impact a variety of functions such as cell proliferation, cell adhesion and bacterial infection of cells.
  • the therapeutic methods of the invention generally comprise administration of a therapeutically effective amount of a GalT-2 inhibitor compound to a subject in need of such treatment, such as a mammal, and particularly a primate such as a human.
  • Treatment methods of the invention also comprise administration of an effective amount of a compound of Formula I as defined herein to a subject, particularly a mammal such as a human in need of such treatment for an indication disclosed herein.
  • Typical subjects include mammals suffering from or susceptible to those conditions discussed above, i.e. proliferative disorders such as vasculopathies, e.g.
  • Atherosclerosis and restenosis such as may occur following an angioplasty procedure; pathologies involving neovascularization; tumor or cyst development, e.g., polycystic kidney disease and post-surgical keloid formation such as may occur on the skin of a patient; inflammatory diseases involving a proinflammatory cytokine such as TNF- ⁇ or interleukin-6 (IL-6); and lipid storage diseases such as Gaucher's disease.
  • IL-6 interleukin-6
  • GalT-2 inhibitor compounds can be employed in the present treatment methods. Simple testing, e.g., in a standard in vitro assay as defined above, can readily identify suitable GalT-2 inhibitor compounds.
  • Preferred GalT-2 inhibitor compounds include those that contain a propanol backbone.
  • Generally preferred for use in the treatment methods of the invention are compounds of the following Formula I:
  • R and R 1 are independently selected from the group consisting of hydrogen and straight-chained or branched C ⁇ -C 6 alkyl with or without a substituent such as amino, hydroxy or mercapto and further wherein R and R 1 may be taken together to form a 5, 6 or 7-membered ring substituent such as pyrrolidino, morpholino, thiomorpholino, piperidino, azacycloheptyl and the like;
  • R 2 is selected from the group consisting of branched or straight-chained C 6 - C 0 alkyl with or without one to three double bonds; and R is selected from the group consisting of straight-chained or branched C 6 -
  • Suitable compounds of Formula I above and other GalT-2 inhibitor compounds can be readily prepared by known procedures or can be obtained from commercial sources. See, for example, Abe, A. et al., (1992) J. Biochem. 111:191- 196; Inokuchi, J.
  • a treatment compound can be administered to a subject in any of several ways.
  • a GalT-2 inhibitor compound can be administered as a prophylactic to prevent the onset of or reduce the severity of a targeted condition.
  • a GalT-2 inhibitor compound can be administered during the course of a targeted condition.
  • a treatment compound can be administered to a subject, either alone or in combination with one or more therapeutic agents, as a pharmaceutical composition in mixture with conventional excipient, i.e. pharmaceutically acceptable organic or inorganic carrier substances suitable for parenteral, enteral or intranasal application which do not deleteriously react with the active compounds and are not deleterious to the recipient thereof.
  • conventional excipient i.e. pharmaceutically acceptable organic or inorganic carrier substances suitable for parenteral, enteral or intranasal application which do not deleteriously react with the active compounds and are not deleterious to the recipient thereof.
  • Suitable pharmaceutically acceptable carriers include but are not limited to water, salt solutions, alcohol, vegetable oils, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oil, fatty acid monoglycerides and diglycerides, petroethral fatty acid esters, hydroxymethyl-cellulose, polyvinylpyrrolidone, etc.
  • the pharmaceutical preparations can be sterilized and if desired mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously react with the active compounds.
  • auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously react with the active compounds.
  • compositions may be prepared for use in parenteral administration, particularly in the form of liquid solutions or suspensions; for oral administration, particularly in the form of tablets or capsules; intranasally, particularly in the form of powders, nasal drops, or aerosols; vaginally; topically e.g. in the form of a cream; rectally e.g. as a suppository; etc.
  • the pharmaceutical agents may be conveniently administered in unit dosage form and may be prepared by any of the methods well known in the pharmaceutical arts, e.g., as described in Remington 's Pharmaceutical Sciences (Mack Pub. Co., Easton, PA, 1980).
  • Formulations for parenteral administration may contain as common excipients such as sterile water or saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, hydrogenated naphthalenes and the like.
  • biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be useful excipients to control the release of certain GalT-2 inhibitor compounds.
  • parenteral delivery systems include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes.
  • Formulations for inhalation administration contain as excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or oily solutions for administration in the form of nasal drops, or as a gel to be applied intranasally.
  • Formulations for parenteral administration may also include glycocholate for buccal administration, methoxysalicylate for rectal administration, or citric acid for vaginal administration.
  • Other delivery systems will administer the therapeutic agent(s) directly at a surgical site, e.g. after balloon angioplasty a GalT-2 inhibitor compound may be administered by use of stents.
  • a GalT-2 inhibitor compound can be employed in the present treatment methods as the sole active pharmaceutical agent or can be used in combination with other active ingredients, e.g., probucol, known antioxidants (e.g. Vitamin C or E) or other compounds.
  • concentration of one or more treatment compounds in a therapeutic composition will vary depending upon a number of factors, including the dosage of the GalT-2 inhibitor compound to be administered, the chemical characteristics (e.g., hydrophobicity) of the composition employed, and the intended mode and route of administration.
  • one or more than one of the GalT-2 inhibitor compounds may be provided in an aqueous physiological buffer solution containing about 0.1 to 10% w/v of a compound for parenteral administration.
  • Suitable dose ranges may include from about l ⁇ g/kg to about lOOmg/kg of body weight per day.
  • Therapeutic compounds of the invention are suitably administered in a protonated and water-soluble form, e.g., as a pharmaceutically acceptable salt, typically an acid addition salt such as an inorganic acid addition salt, e.g., a hydrochloride, sulfate, or phosphate salt, or as an organic acid addition salt such as an acetate, maleate, fumarate, tartrate, or citrate salt.
  • a pharmaceutically acceptable salt typically an acid addition salt such as an inorganic acid addition salt, e.g., a hydrochloride, sulfate, or phosphate salt, or as an organic acid addition salt such as an acetate, maleate, fumarate, tartrate, or citrate salt.
  • Pharmaceutically acceptable salts of therapeutic compounds of the invention also can include metal salts, particularly alkali metal salts such as a sodium salt or potassium salt; alkaline earth metal salts such as a magnesium or calcium salt; ammonium salts such an ammonium or tetramethyl ammonium salt; or an amino acid addition salts such as a lysine, glycine, or phenylalanine salt.
  • metal salts particularly alkali metal salts such as a sodium salt or potassium salt
  • alkaline earth metal salts such as a magnesium or calcium salt
  • ammonium salts such an ammonium or tetramethyl ammonium salt
  • an amino acid addition salts such as a lysine, glycine, or phenylalanine salt.
  • Preferred GalT-2 inhibitor compounds exhibit significant activity in a standard cell proliferation assays.
  • the GalT-2 inhibitor compound inhibits cell proliferation by at least 15 or 25%, preferably at least 50%, relative to a suitable control assay. In such an assay, between about 0.1 to lOO ⁇ M, preferably between about 1 to 50 ⁇ M of a desired GalT-2 inhibitor compound is used.
  • Exemplary cell proliferation assays include counting viable cells and monitoring activity of specified citric acid cycle enzymes such as lactate dehydrogenase.
  • a preferred assay measures incorporation of one or more detectably-labeled nucleosides into DNA, e.g., by: a) culturing suitable cells in medium and adding 1) a candidate GalT-2 inhibitor compound and 2) a radiolabeled nucleoside such as H- thymidine typically in an amount between about 0.1 to lOO ⁇ Ci; b) incubating the cells, e.g., for about 6-24 hours, and typically followed by washing; and c) measuring incorporation of the radiolabeled nucleoside into DNA over that time relative to a control culture that is prepared and incubated under the same conditions as the assay culture but does not include the potential GalT-2 inhibitor compound.
  • TCA trichloroacetic acid
  • references herein to a "standard in vitro cell proliferation assay” or other similar phrase refer to an assay that includes the above steps a) through c).
  • a cell proliferation assay uses aortic smooth muscle cells (ASMCs), particularly those obtained from a human, cow or a rabbit.
  • ASMCs aortic smooth muscle cells
  • a suitable protocol involves preparing ASMCs according to standard methods and culturing same in microtifre plates in a suitable medium such as Ham's F-10.
  • a desired GalT-2 inhibitor compound is then diluted in the medium, preferably to a final concentration of between about 1 to lOO ⁇ g, more preferably between about 1 to 50 ⁇ g per ml of medium or less followed by an incubation period of between about 1-5 days, preferably about 1 day or less.
  • a standard cell proliferation can be conducted, e.g., incorporation of tritiated thymidine or lactate dehydrogenase assay as mentioned above.
  • the assays are preferably conducted in triplicate with a variation of between 5% to 10%. See e.g., Ross, R. J. Cell. Biol. (1971) 50:172; Chatterjee, S. et al. (1982) Eur. J. Biochem. 120:435; Bergmeyer, H.V. In Principles of Enzymatic Analysis. (1978) Verlag Chemie, NY.
  • preferred GalT-2 inhibitor compounds exhibit significant activity in a conventional cell adhesion assay.
  • the GalT-2 inhibitor compound inhibits cell adhesion by at least 25%, preferably at least 50% or more relative to a suitable control assay.
  • a suitable control assay between about 0.1 to lOO ⁇ M, preferably between about 1 to 50 ⁇ M of a desired GalT-2 inhibitor compound is used.
  • a preferred cell adhesion assay includes the following steps: a) labeling a first population of immune cells, preferably certain leukocytes, with a detectable label which can be a chromatic, radioactive, luminescent (e.g., fluorescent, or phosphorescent), or enzymatic label capable of producing a detectable label, b) contacting the first population of cells with a second population of endothelial cells detectably-labeled, e.g., with a chromatic, radioactive, luminescent (e.g., fluorescent or phosphorescent), or enzymatic label preferably different from the label employed in step a); and c) detecting any adhesion between the first and second population of cells.
  • a detectable label which can be a chromatic, radioactive, luminescent (e.g., fluorescent, or phosphorescent), or enzymatic label capable of producing a detectable label
  • a second population of endothelial cells detectably-labeled,
  • references herein to a "standard in vitro cell adhesion assay” or other similar phrase refer to an assay that includes the above steps a) through c).
  • the detection in step c) can be achieved by a variety of methods such as microscopy, particularly confocal microscopy and fluorescence-based photomicroscopy involving FACS; automated cell sorting techniques, immunological methods such as ELISA and RIA; and scintillation counting. See examples below for disclosure relating to preferred cell adhesion assays.
  • a preferred in vitro cell adhesion assay measures polymorphonuclear leukocytes (PMNs and/or myocytes) or platelets and increased endothelial cell adhesion before, during or after contact with a desired GalT-2 inhibitor compound.
  • PMNs and/or myocytes polymorphonuclear leukocytes
  • platelets increased endothelial cell adhesion before, during or after contact with a desired GalT-2 inhibitor compound.
  • the PMNS or myocytes can be collected and purified according to standard methods detailed below.
  • the PMNs or myocytes are then labeled by incubation with a suitable fluorescent dye such as fluorescent Cell Tracker dye (e.g., green) or Calcein-AM.
  • a suitable fluorescent dye such as fluorescent Cell Tracker dye (e.g., green) or Calcein-AM.
  • an endothelial cell monolayer prepared in accordance with standard cell culture methods on a suitable substrate such as a slide or a sterilized plastic petri dish is contacted by the GalT-2 inhibitor compound and labeled with another fluorescent dye such as fluorescent Cell Tracker dye (e.g., orange).
  • the PMNs or myocytes and endothelial cells are then incubated for between about 10 minutes to a few hours, preferably about 30 minutes at 37°C.
  • Non-adherent cells are then washed away from the slide with a physiologically acceptable buffer such as phosphate-buffered saline (PBS).
  • Adhering cells are then quantitated by standard methods such as by use of a fluorescence plate reader.
  • the number of adherent cells on the slide can be quantitated in several ways including expressing the number of PMN/m ⁇ T on the endothelial cell monolayer.
  • the adhering cells can be quantitated by inspection following photomicroscopy visualized and photographed by microscopy. Cell adherence is then evaluated by inspection of the photomicrograph. See the examples which follow.
  • GalT-2 assays conducted with the ASMCs and performed in accordance with previously described methods. See e.g., Chatterjee, S., and Castiglione, E. (1987) Biochem. Biophys. Acta, 923: 136; and Chatterjee, (1991) S. Biochem. Biophys. Res Comm., 181:554.
  • Suitable in vitro cell adhesion assays include immunological detection of adhesion molecules on PMNs using specified antibodies, particularly monoclonals, capable of specifically binding the adhesion molecules.
  • a particularly preferred assay involves flow cytometry.
  • ICAM-1 intracellular adhesion molecule 1
  • Mac-1 CDl lb/CD 18
  • LFA-1 selectin
  • Another preferred assay of the invention includes the following steps a) through d): a) culturing a population of LacCer-responsive cells preferably to confluency in lipoprotein-deficient serum medium, e.g., about lmg lipoprotein- deficient serum/protein/ml of medium or less; b) harvesting the cells preferably in a suitable dispersive buffer, e.g., cacodylate buffer; c) incubating the harvested cells preferably with a detectably labeled molecule such as a detectably-labeled nucleoside diphosphate sugar donor such as [ 14 C]-UDP-galactose typcially in an amount between about 0.1 to lOO ⁇ Ci; and d) measuring LacCer formation as indicative of the activity of the GalT-2 enzyme.
  • a detectably labeled molecule such as a detectably-labeled nucleoside diphosphate sugar donor such as [ 14 C]-UDP-galactose typcially
  • the assays generally described above will use known LacCer-responsive cells and will be cultured in a medium suitable for maintaining those cells in the assay, e.g., Eagles's minimum essential medium (HMEM) or Ham's F-10 medium.
  • HMEM Eagles's minimum essential medium
  • Further preferred GalT-2 inhibitor compounds include those that exhibit at least a 2- to 5-fold greater inhibition of GalT-2 relative to GlcT-1 as measured by conventional GlcT-1 and GalT-2 enzyme assays. More preferred are those GalT-2 inhibitor compounds that exhibit at least about 5- to 10-fold greater inhibition of GalT-2 relative to inhibition of GlcT-1, even more preferably at least about 10- to 50- fold. Methods for measuring GlcT-1 have been reported. See e.g., Carson, K., and Ganem, B. supra; Shukla, A. and Radin, N.S. J. Lipid. Res. 32:713.
  • GalT-2 inhibitor compounds include those that are capable of specifically inhibiting the GalT-2 enzyme. That is, the identified GalT-2 inhibitor compound is a relatively poor inhibitor of other enzymes relating to GSLs such as hydroxyceramide galactosyltransferase, glucocerebroside glucosidase, and particularly GlcT-1. Significantly, the GalT-2 inhibitor compound should avoid undesired pharmacological effects that could arise from non-selective inhibition of other GSL-related enzymes. Exemplary of such preferred GalT-2 inhibitor compounds are those which are GalT-2 transition state mimics. The in vivo assays of the invention are particularly useful for subsequent evaluation of GalT-2 inhibitor compounds exhibiting suitable activity in an in vitro assay.
  • a rabbit model of restenosis accompanying an invasive surgical procedure such as balloon angioplasty is preferred.
  • One suitable protocol involves administering to the rabbit a suitable vehicle or vehicle combined with one or more GalT-2 inhibitor compounds of interest.
  • the amount of the GalT-2 inhibitor compound administered will vary depending on several parameters including the extent of damage associated with the surgical procedure of interest.
  • the rabbit will typically receive a candidate GalT-2 inhibitor compound in a dose (e.g., i.m. or i.p.) of between about 0.5 to 100, preferably 1 to 20 and more preferably about lOmg/kg body weight of the rabbit.
  • a preferred dosage schedule provides for administration of a GalT-2 inhibitor compound starting 24 hurs prior to conducting an invasive surgical procedure, and then continuing administration of the GalT-2 inhibitor compound for 15 days following the surgical procedure.
  • daily injections of the GalT-2 inhibitor compound may be made for about 2 to 12 weeks following the invasive surgical procedure.
  • Daily injections, e.g., i.m. or i.p., of the GalT-2 inhibitor compound are generally preferred.
  • the rabbits are euthanized and a vessel removed for examination, preferably the aorta. The vessel is then fixed with formalin and analyzed for proliferation of vascular endothelia, media and advantitia using standard histological procedures.
  • invasive surgical procedure means a medical or veterinary technique associated with significant damage to the endothelium of a vessel impacting, e.g., an organ such as the heart, liver or the kidney, or a limb.
  • a vessel comprises the aorta, coronary vessel, femoral and iliac arteries and veins.
  • the invasive surgical procedure can be associated with techniques involving, e.g., cardiac surgery, abdominothoracic surgery, arterial surgery, deployment of an implementation (e.g., a vascular stent or catheter), or endarterectromy.
  • a preferred invasive surgical procedure is angioplasty, particularly balloon angioplasty.
  • the invasive surgical procedure is performed on a mammal such as a primate, particularly a human, rodent or a rabbit, or a domesticated animal such as a pig, dog or a cat.
  • the present invention includes methods of detecting and analyzing GalT-2 inhibitor compounds with therapeutic capacity to treat or prevent any of the above-mentioned diseases, post-surgical disorders, or bacterial infections modulated by LacCer.
  • a disease, post-surgical disorder or bacterial infection is suitably considered as being modulated by LacCer if afflicted cells or tissue exhibit GalT-2 activity about 2- to 50-fold, typically about 2- to 10-fold, and more typically about 2- to 5-fold higher than that of control (unafflicted) cells or tissue.
  • the GalT-2 activity can be measured by methods referenced herein. Without being bound by theory, it appears that increased GalT-2 activity produces substantial amounts of LacCer.
  • LacCer is believed to enhance the onset of or contribute to the severity of the specified diseases, post-surgical disorders and bacterial infections.
  • GalT-2 levels in renal tissue afflicted by polycystic kidney disease exhibit a 3 -fold elevation in GalT-2 activity and LacCer levels as compared to control renal tissue.
  • polycystic kidney disease is one example of a disease impacted by LacCer. See Chatterjee, S. et al. J. Lipid Res. (1996) 37:1334.
  • the novel LacCer-related steps disclosed herein have been found to relate changes in GalT-2 activity to cell proliferation or adhesion in LacCer- responsive cells. It has been determined that the LacCer-related steps can be grouped into those inhibiting cell proliferation and those effecting cell adhesion.
  • the LacCer- related steps have been found to include a variety of identified molecules such as specified enzymes, cytosolic factors, nuclear factors, radical species and adhesion proteins. More particular examples of such molecules in the LacCer-related biochemical steps include GTP-binding proteins, kinases, cytosolic factors, nuclear factors, transcription factors, and oxygen species, particularly reactive oxygen species (sometimes referred to herein as "ROS" or "ROM").
  • Detection methods of the invention are formatted to include one or more steps associated with LacCer-related pathways. More particularly, the detection methods include specific steps that measure the activity of molecules which act to modulate cell proliferation or adhesion. In some cases, a particular molecule will act to inhibit both cell proliferation and adhesion through a LacCer-related pathway.
  • a LacCer-responsive cell can be an immortalized cell line or primary culture of cells (e.g., obtained form a tissue or organ) that manifests a change in one or more specific cell molecules or functions such as proliferation or adhesion, following contact with a suitable amount of LacCer. More specifically, one or a combination of strategies can identify a LacCer- responsive mammalian cell. For example, in one approach, about 1 x 10 5 cells are seeded in petri dishes in suitable growth medium.
  • a desired tissue or organ is obtained from an animal and dispersed according to standard methods (e.g., by sonication, mechanical agitation, and/or exposure to dispersing agents known in the field, e.g., detergents and proteases). After one or a few days, the growth medium is removed from the petri dish and the cells washed with phosphate- buffered saline. The cells are then primed in a suitable medium for about 1 to 5 hours at which point LacCer is added to culture. The amount of LacCer added will depend on several parameters such as the particular cell or tissue type being tested.
  • the LacCer will be added to the culture at a concentration of between about l ⁇ g to lmg, preferably between about l ⁇ g to 500 ⁇ g, and more preferably between about l ⁇ g to 50 ⁇ g per ml of culture medium.
  • the medium is removed and the cells lysed in an appropriate lysis buffer such as those described in detail below. The cells are then assayed according to any of the methods described herein for response to the added LacCer.
  • LacCer-responsive mammalian cells include those cells associated with smooth muscle cells, e.g., cells associated with the vasculature of an organ or limb, particularly heart or kidney cells. More particularly, ASMCs
  • H-ASMCs sometimes referred to herein as H-ASMCs to denote human origin
  • endothelial cells also preferred are certain immune cells such as white blood cells, particularly PMNs and monocytes.
  • Preferred GalT-2 inhibitor compounds also include those that exhibit good capacity to modulate one or more specified molecules in a LacCer-related step following exposure to LacCer.
  • Particularly preferred compounds exhibit at least 20%, preferably at least 50% and more preferably at least 90% or more of a decrease in the activity of the molecule (relative to a suitable control assay) at a concentration of between about 0.1 to lOO ⁇ g/ml, preferably between about 1 to lO ⁇ g/ml in an in vitro detection assay.
  • the activity of the molecules can decrease in any of several readily detectable ways including altered synthesis, degradation or storage; protein modification, e.g., phosphorylation, or through an allosteric effect as with certain enzymes.
  • preferred GalT-2 inhibitor compounds include those that exhibit good activity in an enzyme assay as described below.
  • an IC 50 in such an assay is about 20 ⁇ M or less, more preferably an IC 50 about 1 ⁇ M or less.
  • a control experiment is generally tailored for use in a particular assay. For example, most control experiments involve subjecting a test sample (e.g., a population of LacCer-responsive cells or lysate thereof) to medium, saline, buffer or water instead of a potential GalT-2 inhibitor compound in parallel to the cells receiving an amount of test compound. A desired assay is then conducted in accordance with the present methods. Specific examples of suitable control experiments are described below.
  • a test sample e.g., a population of LacCer-responsive cells or lysate thereof
  • medium, saline, buffer or water instead of a potential GalT-2 inhibitor compound
  • the present detection methods also can be used to identify GalT-2 inhibitor compounds obtained from biological sources, including specified growth factors, cytokines, and lipoproteins that modulate GalT-2 activity.
  • the present detection methods further include assays v/hich measure the activity of specified molecules in LacCer-related biochemical steps.
  • the measurements can be conducted by standard laboratory manipulations such as chemiluminescence tests, thin layer chromatography (TLC) separations, nucleic acid isolation and purification, SDS-PAGE gel electrophoresis, autoradiography, scintillation counting, densitometery, Northern and Western Blot hybridization, and im unoassays (e.g., R ⁇ A and ELISA tests).
  • TLC thin layer chromatography
  • SDS-PAGE gel electrophoresis autoradiography
  • scintillation counting e.g., densitometery
  • Northern and Western Blot hybridization e.g., R ⁇ A and ELISA tests.
  • the present in vitro assays measure the activity of certain enzymes in LacCer-responsive cells.
  • the activity of the enzymes has been found to be modulated following exposure of the cells to LacCer and/or a specified GalT-2 inhibitor compound such as PDMP, oxidized lipoprotein (ox-LDL), nerve growth factor (NGF), platelet derived growth factor (PDGF), epidermal growth factor (EGF), and tumor necrosis factor- ⁇ (TNF- ⁇ ).
  • a specified GalT-2 inhibitor compound such as PDMP, oxidized lipoprotein (ox-LDL), nerve growth factor (NGF), platelet derived growth factor (PDGF), epidermal growth factor (EGF), and tumor necrosis factor- ⁇ (TNF- ⁇ ).
  • D-PDMP has been found to reduce the activity of a variety of such enzymes including specified redox enzymes, GTP -binding proteins, and kinases discussed below.
  • one particular in vitro assay measures the activity of an oxidase capable of synthesizing an oxygen species, particularly a ROS such as superoxide.
  • a particularly preferred enzyme is NADPH oxidase.
  • the activity of the NADPH oxidase can be assayed by standard methods including fractionating the enzyme from cell components and then measuring the activity by enzyme assay such as those employing a standard chemiluminescence method.
  • the NADPH oxidase can be assayed by measuring superoxide production in intact cells.
  • the measurement is conducted in the presence of a mitochondrial poison such as KCN, an inhibitor of NADH oxidase.
  • the activity of the NADPH oxidase can be assayed in intact LacCer-responsive cells by measuring superoxide production.
  • the superoxide measurement can be performed in several ways including incubating the cells with a photosensitive polycyclic organic compound (e.g., an acridylium compound). Reduction of the polycyclic compound by superoxide causes light emission that can be detected by a standard photon counter. Preferred methods of measuring the NADPH oxidase activity are described in Bhunia, A.K. et al. (1997) J. Biol. Chem. 275:15642.
  • Additional in vitro assays are provided which measure one or more enzymes that have been found to be modulated by LacCer and GalT-2 inhibitor compounds disclosed herein.
  • the enzymes include Ras-GTP -binding protein, Raf-1, mitogen activated protein (MAP) kinase (MEK-2), and other mitogen activated protein kinases such as p44 MAPK.
  • MAP mitogen activated protein
  • MEK-2 mitogen activated protein kinases
  • p44 MAPK mitogen activated protein kinases
  • incorporation of a nucleoside triphosphate, particularly a cyclic nucleoside triphosphate such as guanidine nucleoside triphosphate (GTP) into an oncogene protein such as the ras protein (i.e. ras-GTP loading) by the ras-GTP - binding protein can be measured by a number of distinct approaches including direct detection of nucleoside triphosphate (e.g., GTP) incorporation into Ras.
  • GTP guanidine nucleoside triphosphate
  • LacCer-responsive cells are metabolically labeled with radioactive orthophosphate (e.g., 32 P-labeled) to detectably-label the GTP inside the cells.
  • the labeled cells are incubated with LacCer followed by a GalT-2 inhibitor compound and then washed and lysed in a suitable lysis buffer such as RIPA (see below). Subsequently, the cell lysate is separated on suitable TLC plates. The TLC plates are exposed to X-ray film and then subjected to densitometery, if desired, to quantitate incorporation of the GTP into the Ras protein.
  • a preferred method for detecting ras- GTP loading has been disclosed in Chatterjee, S. et al., (1997) Glycobiology, 7:703. Methods are also provided for measuring the activity of the Raf-1 and Mek-2 enzymes.
  • the LacCer-responsive cells are incubated with LacCer and a potential GalT-2 inhibitor compound, washed, and then harvested after about 1 to 60 minutes, preferably 1 to 10 minutes or less, after exposure to the LacCer.
  • Whole cell lysates are prepared and then subjected to standard SDS-PAGE gel electrophoresis. The gels are transferred to a suitable membrane support and then probed with anti-RAF-1 or anti-MEK antibody in accordance with conventional
  • DNA binding proteins e.g., transcription factors such as c-fos, or the nuclear factor kB
  • DNA binding protein (NF-kB). These DNA binding proteins have been surprisingly found to be modulated by LacCer and GalT-2 inhibitor compound. The DNA binding proteins can be assayed by a number of conventional approaches.
  • the activity of the NF-kB DNA binding protein can be measured by a standard polyacrylamide gel mobility shift assay.
  • the gel assay is performed after contacting LacCer-responsive cells with LacCer followed by a potential GalT-2 inhibitor compound.
  • a cell lysate is prepared from the LacCer-responsive cells which is then contacted with an oligonucleotide sequence comprising (or consisting of) a recognized NF-kB DNA binding sequence.
  • the reaction mixture is then incubated for a time sufficient to allow the NF-kB protein and the DNA binding sequence to form a specific binding complex.
  • the specific binding complex is then separated on an SDS-PAGE polyacrylamide gel which is subsequently dried and exposed to X-ray film.
  • PCNA proliferating cell nuclear antigen
  • the cultured cells are incubated with LacCer followed by a GalT-2 inhibitor compound and then washed with a suitable buffer.
  • PCNA in the cultured cells can be detected (and quantified if desired) by using a monoclonal antibody that is capable of specifically binding the PCNA (e.g., PC10 antibody). See Sasaki, K., et al. (1993) Cytometry 14:876-882.
  • the PCNA then can be detected in the cells by a variety of immunological methods including flow cytometery or imunohistochemical visualization of fixed cell sections.
  • glycosphingolipids and all other chemicals were purchased from Sigma Chemical Company, (St. Louis, U.S.A). The purity of glycosphingolipids (>99%) was assessed by HPLC and or HPTLC.
  • ICAM-1 antibody and SOD antibody were purchased from SantaCruz Biotechnology, CA. Cell Tracker dye green and cell Tracker dye orange were obtained Molecular Probes Inc., CA. DPI was from Calbiochem. USA.
  • Hybridoma Eahy 926 derived from human umbilical vein endothelial cells (HUVEC; Clonetech) and human epithelial cell line A549, was a kind gift from Dr. Roger Harrison (University of Bath, UK). These cells were cultured in plastic flasks in RPMI 1640 medium (Gibco BRL, Gaithersburg, MD) containing 10% fetal calf serum, penicillin (lOOU/ni) and streptomycin (O.lmg/mL) (Gibco), passaged at confluence using 0.05% trypsin and 0.53 mmoI/mL EDTA (Gibco), and then grown to confluence in 24 well plates.
  • This suspension was consistently found to be comprised of greater than 95% PMNs by microscopic morphology following modified Wright-Giemsa staining (Diff-Quik Stain Set, Baxter, Miami, FL). 3. Vehicle for Glycospingohpids.
  • Stock solution of LacCer and other glycosphingolipids were prepared in DMSO and added to culture medium to achieve the desired concentrations of LacCer. Cells incubated with 0.01% DMSO served as a control.
  • DPI stock solutions were prepared in DMSO and stored at -20°C until use.
  • Aqueous solutions of NAC or allopurinol were prepared either in medium or in buffer.
  • Lucigenin an acridylium compound (Sigma) that emits light on reduction and interaction with O 2 " was used to measure O 2 " production by chemiluminescence as described above.
  • Confluently grown endothelial cells were harvested and O " was measured in intact endothelial cell suspension using a 96-well plate containing dark adapted lucigenin (500 ⁇ M) in balanced salt solution.
  • the viability of the suspended cells as determined by trypan blue exclusion principle was >90%. LacCer was added to it as a stimulant and photon emission was measured every 20 seconds for 10mm in a scintillation counter (Packard TOP counter, USA).
  • the GSL solutions (dissolved in DMSO) were added to cells to reach a final concentration of DMSO of 0.01 %.
  • NF-kB DNA Binding Assay Gel mobility shift assays of NF-kB DNA binding assays were performed as described (Yin et al. (1996) J. Biol. Chem., 271 : 17974). Briefly, 20 ⁇ l reaction mixture contained 8- 1 O ⁇ g of nuclear protein plus a [ 32 P] -labeled oligonucleotide probe containing NF-kB binding site (5'- AGTTGAGGGGACTTCCCAGGC-3' SEQ ID No. 1) (Lenardo, M.J. and D.
  • ICAM-1 Expression Assay A modified ELISA in 96-well plates determined quantitative measurement of the expression of ICAM-1 on the surface of the endothelial cell monolayer. After the treatment of endothelial cell monolayers with or without LacCer, the cells were fixed with 3.7% formaldehyde (pH 7.4) containing 0.1M Lysine monohydrochloride and 0.01M sodium m-periodate for 20 minutes at 4°C and then blocked with PBS containing 1% BSA and 0.1M glycine overnight at 4°C. The fixed monolayer was then probed with mouse monoclonal anti- human ICAM-1 antibody for 1 hour at 37°C.
  • ICAM-1 expression was assessed qualitatively by immunofluorescence staining using fluorescent isothiocyanate (FITC) -conjugated IgG after fixing with or without LacCer incubated cells with 3.7% formaldehyde.
  • FITC fluorescent isothiocyanate
  • Eahy endothelial cells in 96-well plate were infected with virus containing CuZn-SOD gene in multiplicities of infection (MCI) ranging from 0 to 75.
  • MCI multiplicities of infection
  • Another plate of endothelial cells were infected with control virus (ad- ⁇ -gal) of similar dilution. Twenty-four hours later, the cells were washed with PBS to remove uninfected virus particles. The endothelial cells were then incubated for an additional 48 hours in fresh medium. Next, the cells were fixed with 3.7% formaldehyde in PBS.
  • the expression of intracellular SOD by the endothelial cells was measured by ELISA after permeabilization with 0.2% Triton X- 100 in PBS for 20mm.
  • the permeabilized endothelial cells were probed with monoclonal anti-human SOD antibody and then with FITC- conjugated goat anti-mouse IgG.
  • FITC fluorescence was measured employing a fluorescence plate reader (CytoFluor 2300) at 480nm (excitation) and 530nm (emission).
  • SOD activities in cell extracts were measured by the method of McCord and Fridovich (McCord, J.M. and I. Fridovich (1969) J. Biol. Chem., 244:6049).
  • PMNs polymorphonuclear leukocytes
  • human blood was collected in lOU/ml heparin from laboratory volunteers. Blood was centrifuged at 1300 X g for 10mm at 4°C. The white blood cell layer was removed and layered over cold Accu-prep gradient (Accurate Chemical and Scientific Corp., (Westbury, NY), and centrifuged at 600 X g for 30mm for the separation of leukocytes. The red blood cell (RBC)/PMN layer was resuspended in RBC lysing buffer (Sigma St. Louis).
  • DCFH- DA 2'J'-dichlorofluorescein diacetate (Molecular Probes, Eugene, OR) was used to determine the net cellular generation of ROMs by PMNs. 2 x 10 5 PMNs/well were incubated in 96 well-plates with/without various concentrations of N-acetylcysteine (NAC, a cell permeant, relatively nonselective antioxidant) (Sigma) or diphenyleneiodonium chloride (DPI, a NADPH oxidase inhibitor) (Sigma) for 20 minutes at 37°C, then DCFH-DA (final concentration 5 ⁇ mol/L).
  • NAC N-acetylcysteine
  • DPI diphenyleneiodonium chloride
  • PMN-Endothelial Adhesion Assay PMNs were labeled fluorescently by incubation with 5 ⁇ mol/L Calcein-AM (Molecular Probes) in calcium-free PBS for 20 minutes at 37°C, washed and resuspended in Hanks' balanced salt solution (Gibco) containing 0.2% bovine serum albumin (BSA). These PMNs were then treated with LacCer (or vehicle, 0.05% DMSO), washed 3 times with PBS and then plated on the unstimulated ec monolayers, which had been grown to confluence in 24-well plates.
  • Calcein-AM Molecular Probes
  • BSA bovine serum albumin
  • the PMNs (600 ⁇ L of 2 x 10 6 /mL/ well) then were incubated with the ecs for 30 minutes at 37°C. Non-adherent PMNs were then removed by gentle washing 3 times with PBS. The residual adherent PMNs (and ecs) were then lysed using 4mmol/L Zwittergent (Calbiochem, La Jolla, CA) and the plates read on a fluorescence plate reader (Millipore) at EX 480nm/EM 530nm. The number of adherent PMNs were expressed as number of PMN/mm ⁇ ec monolayer, based on the mean fluorescent intensity of each PMN, as determined from a standard curve.
  • LacCer, ceramide and GlcCer (all from Sigma) were prepared as described in examples below, and used as putative agonists for the stimulation of PMN adhesion molecule expression, assayed by FACScan.
  • PMNs were incubated with the following specific inhibitors for 20 minutes at 37°C, and then stimulated with lOOnmol/L LacCer for 20 minutes: NAC (a cell permeable, relatively nonselective antioxidant); DPI (a neutrophil NADPH oxidase inhibitor); or
  • BAPTA/AM an intracellular calcium buffer
  • Genistein (Calbiochem), staurosporine (Calbiochem), or quinacrine dihydrochloride (ICN, Aurora, Ohio) was used for evaluating the possible involvement of tyrosine kinase, protein kinase C (PKC), or phospholipase A2 (PLA2), respectively, in LacCer-induced Mac-1 upregulation in hpMN.
  • PDC protein kinase C
  • PLA2 phospholipase A2
  • WEB 2086 (a specific PAF-receptor antagonist) (Boehringer Ingelheim, Ridgefield, CT) was used to determine the possible involvement of PAF, which is one of the metabolites metabolically downstream from PLA2 which can upregulate PMN Mac-1.
  • PAF PAF-receptor antagonist
  • the ec monolayers in the half of each 24 well-plate were first fixed with 2% buffered formalin for 5 minutes and then washed 3 times with PBS. The ecs on the other half of the plate were left unfixed, but similarly washed before use for the adhesion assay.
  • PMNs were first incubated with increasing doses of quinacrine for 20 minutes, then stimulated with lOOnmol/L of LacCer for 20 minutes, followed by washing 3 times, and then plated onto formalin-prefixed ec monolayers in the presence of each concentration of quinacrine to eliminate a possible direct effect of quinacrine on the ecs.
  • PMNs were first incubated with LacCer for 20 minutes at 37°C,washed 3 times, incubated with m-anti- CD 18 F(ab') 2 (mHm23, a generous gift from Dr. J. Hildreth, The Johns Hopkins University, Baltimore, MD) for 15 minutes at 37°C, and then plated onto the ec monolayers without washing.
  • Example 1- D-PDMP inhibits TNF- ⁇ Induced ICAM-1 Expression
  • the level of GlcCer and LacCer was found to decrease significantly in rabbits receiving D-PDMP. Concomitantly, the activity of GaIT-2 was also decreased ⁇ 1J- fold (see Table 1 below). Thus, D-PDMP inhibited GalT-2 activity and decreased the level of LacCer in rabbit thoracic aorta. The results also indicate that rabbit balloon angioplasty is a feasible model to study in vivo effects of D-PDMP or other GalT-2 inhibitor compounds on stenosis.
  • rabbits were given intramuscular injections of vehicle alone or lOmg D-PDMP/kg in vehicle. Daily injections of D-PDMP continued for six weeks after balloon angioplasty.
  • the rabbits were euthanized by CO 2 asphyxia and the aorta was surgically removed. The site of balloon angioplasty was marked and the tissue was fixed with 10% formalin in phosphate buffered saline. The samples were prepared for histologic examination and photographed with the use of a Roche digital imaging system.
  • Fig. 2A shows a section through the aorta of a control rabbit given D-PDMP for six weeks. Note that the endothelium, media and adventitia are clearly visible. Following balloon angioplasty and injection of vehicle alone (Fig. 2B), a marked intimal proliferation and expanded IEL characteristic of endothelial damage was observed. In sharp contrast (Fig. 2C), when rabbits were given injections of D-PDMP following balloon angioplasty, the endothelium had regenerated but some IEL expansion was still observed. The most remarkable finding was the complete absence of intimal proliferation at the site of balloon angioplasty of this rabbit.
  • the data in Table 1, above demonstrates that D-PDMP can reduce the level of glucosylceramide and LacCer and decrease the activity of GalT-2 in the aorta of balloon angioplasty rabbits, compared to control.
  • Figs. 2 A-C show photomicrographs of hematoxylin eosin-stained rabbit aorta sections following balloon angioplasty and treatment with D-PDMP, specifically: section of aorta from control rabbit (without balloon angioplasty) injected with D- PDMP is depicted in Fig. 2A; section of aorta from balloon angioplasty rabbit (without treatment of D-PDMP) is depicted in Fig. 2B; and section of aorta from balloon angioplasty rabbit treatment with D-PDMP is depicted in Fig. 2C.
  • LacCer stimulated the generation of O " in a concentration (Fig. 3 A) and time (Fig. 3B) dependent manner in intact Eahy 926 cells, as measured by lucigenin chemiluminescence. Other GSLs and their constituents did not stimulate the generation of O 2 " in Eahy 926 cells. The maximum generation of O 2 - (2Jnmol/mm/mg protein) was observed at 5 ⁇ M concentration of LacCer after 5 minutes.
  • Figure 3A-B shows that LacCer stimulates superoxide generation in Eahy 926 cells. Confluently grown endothelial cell monolayers were harvested and suspended in balanced salt solution.
  • Fig. 3 A shows the effect of various concentrations of LacCer on the rate of generation of superoxide in endothelial cells. Control (vehicle 0.01% DMSO) (O); LacCer (•).
  • Fig. 3B shows the effect of time of incubation with LacCer on the generation of superoxide. Control (vehicle 0.01% DMSO) (O); LacCer (•).
  • Fig. 3C shows the effect of different glycospingolipids and its constituents (5 ⁇ M) as indicated on the rate of generation of superoxide after 2.5 min.
  • NADPH oxidase activity was measured in membrane preparations using NADPH as a co- factor.
  • Stimulated NADPH dependent oxidase activity was observed in membrane preparations of LacCer treated Eahy 926 cells.
  • At 5 minutes about 3-fold stimulation of NADPH oxidase activity was observed with 5 ⁇ M LacCer as compared to non- stimulated cells membrane preparations (Fig. 4A).
  • FIG. 4A shows plasma membrane preparation of cells incubated with/without LacCer were used for the determination of NADPH oxidase activity as described in the general comments above. NADPH oxidase activity in control cells (O) and 5 ⁇ LacCer treated cells (•) and NADPH oxidase activity in control cells membrane after incubation with lO ⁇ M DPI for 30 minutes ( ⁇ ) and in membranes from cells incubated with lO ⁇ M DPI for 30 minutes + 5 ⁇ M LacCer (shaded triangle).
  • Fig. 4B shows NADH oxidase activity in membrane in control cells (clear blocks) and in cells incubated with LacCer (shaded blocks).
  • LacCer stimulated the binding activity of NF-kB to the consensus oligonucleotide sequence specific for NF-kB (SEQ ED. NO.l). LacCer-induced generation of superoxide can induce expression of NF-kB.
  • Fig.5A-B illustrates effect of LacCer on the expression of NF-kB in Eahy cells. Cells incubated with 5 ⁇ M LacCer and different time intervals nuclear extracts were prepared as described below. Nuclear extracts (lO ⁇ g of protein) were incubated with 32 P-labeled NF-kB binding oligonucleotide probe in reaction mixture, reaction was stopped and run into 6% polyacrylamide gel. The gel was dried and autoradiographed.
  • Intact Eahy 926 cells were incubated with various GSLs (lO ⁇ M) and their constituents. Only LacCer specifically stimulated the expression of intracellular cell adhesion molecule- 1 (ICAM-1) (Fig. 6 A-B). Other GSLs and their constituents or gangliosides (GM 1, GM2, GM3, up to 50 ⁇ M) did not stimulate ICAM-1 expression (Fig. 6A). LacCer did not stimulate measurable expression of other adhesion molecules like VCAM-1 or P-selectin in Eahy 926.
  • IAM-1 intracellular cell adhesion molecule- 1
  • Figs. 6 A-C show effects of various glycosphingolipids on ICAM-1 expression in Eahy 926 cells. More specifically, Fig. 6A shows confluently grown Eahy 926 cells in 96-well plate (lxlO 4 cells/well) were incubated with various glycosphingolipids (5 ⁇ M) for 4 hours. Next, cells were fixed with 3.7% formaldehyde in PBS and ICAM-1 expression was measured by modified ELISA assay using monoclonal ICAM-1 antibody as described in material and methods. Cells incubated with 0.01% DMSO served as a control and treatments ICAM-1 expression data was expressed optical density (O.D)/ well. With respect to the results shown in Fig.
  • endothelial cells were confluently grown in glass chamber slide and incubated with 5 ⁇ M LacCer for 4 hours. Next, cells were fixed in 3.7% formaldehyde and ICAM-1 expression was measured by immunofluorescence using ICAM-1 monoclonal antibody followed by incubation with FITC conjugated anti- mouse IgG and photographed employing fluorescence microscopy.
  • Fig. 6B shows the effect of time and concentration of LacCer on ICAM-1 expression in endothelial cells. ICAM-1 expression was measured as described earlier.
  • LacCer Induced Superoxide Generation and ICAM-1 expression Cells incubated with lO ⁇ M DPI for 30 minutes, failed to respond LacCer induced generation of O 2 " (Fig 7A). Similarly, cells incubated with an antioxidant, N- acetyl cysteine (NAC) (15mM) for 30 minutes, exhibited reduced LacCer induced endogenous levels of O 2 " generation. However, exogenously added SOD did not inhibit the LacCer induced greater level of O 2 " in endothelial cells (Fig. 7A). DPI or the antioxidants NAC or PDTC inhibited LacCer mediated stimulation of ICAM- 1 expression (Fig. 7B). These findings suggest that the NADPH oxidase dependent generation of O 2 " is involved in LacCer induced ICAM-1 expression.
  • LacCer stimulated O 2 " -generation in the endothelial cells in both time and concentration-dependent manner LacCer induced level of O 2 ⁇ was reduced by antioxidant NAC but not by exogenously added superoxide dismutase to cells indicating endogenous generation of O 2 " (Fig. 7A). This observation may be due to the inability of SOD to penetrate the cell membrane but cell permeable NAC reduced the endogenous O 2 " level. LacCer induced O 2 ⁇ endogenously not only in endothelial cells but also induced in smooth muscle cells and in neutrophils. See examples above.
  • Figs. 7 A-B show effects of DPI, NAC and superoxide dismutase on LacCer induced superoxide generation and ICAM-1 expression.
  • Fig. 7 A shows the results of superoxide measured in cells incubated without (O) or with (•) LacCer (5 ⁇ M) and with 15mM NAC ( ⁇ ), cells preincubated with 15mM NAC +5 ⁇ M LacCer (A), cells preincubated with lO ⁇ M DPI (V), cells preincubated with lO ⁇ M DPI for 30 minutes followed by 5 ⁇ M LacCer (V), incubation 200U/ ml SOD ( ⁇ ), 200u/ml SOD+ 5 ⁇ M LacCer ( ⁇ ).
  • Fig. 7 A shows the results of superoxide measured in cells incubated without (O) or with (•) LacCer (5 ⁇ M) and with 15mM NAC ( ⁇ ), cells preincubated with 15mM NAC +5 ⁇ M LacCer (A), cells preincubated
  • FIG. 7B shows the results of ICAM- 1 expression in cells incubated with lO ⁇ M DPI for 30 minutes, lO ⁇ M DPI for 30 minutes + 5 ⁇ M LacCer for 4hours, preincubated with 15mM NAC for 30 minutes, preincubated with 15mM NAC for 30 minutes + 5 ⁇ M LacCer for 4 hours, lOO ⁇ M PDTC for 1 hours, lOO ⁇ M PDTC for 1 hours + 5 ⁇ M LacCer for 4 hours.
  • Fig. 8A-C show adenovirus mediated overexpression of superoxide dismutase and ICAM-1 expression by LacCer in Eahy 926 cells. Overexpression of CuZn-SOD was performed in endothelial cells by adenovirus mediated CuZn-SOD gene transfer as described below.
  • Fig. 8 A shows the results of measurement of SOD expression in endothelial cells by immunofluorescence using monoclonal SOD antibody and FITC- labeled antimouse IgG after fixing cells with 3.7% formaldehyde and permeabilized with 0.2% Triton X-100 in PBS. Control virus ( ⁇ -gal gene only) ( ⁇ ) and Ad-SOD virus ( ⁇ ) infected cells lysate.
  • Fig. 8B shows the results of SOD activity assay in cell lysates of SOD virus (Q) and control virus containing ⁇ -gal gene (shaded square) infected cells.
  • Fig. 8C shows the results of immunofluorescence assay of ICAM- 1 expression in endothelial cells in control virus infected cells (O), control virus infected cells + 5 ⁇ M LacCer for 4 hours (•), Ad-SOD virus infected cells ( ⁇ ) and Ad- SOD virus infected cells + 5 ⁇ M LacCer for 4 hours (T). The data represents mean ⁇ SD of two different experiments.
  • Fig. 9A-D show that LacCer stimulates neutrophil adhesion to Eahy 926 cells.
  • Endothelial cells were confluently grown in glass chamber slides. Then cells were stimulated with 5 ⁇ M LacCer for 4 hours followed by fluorescent (orange color) labeling with cell tracker dye. Neutrophil stained green with a fluorescence probe were coincubated with the endothelial cells for 20 minutes and subjected to confocal microscopy as described below. In some experiments endothelial cells were incubated with either lO ⁇ M DPI or 15mM NAC for 30 minutes prior to incubation with 5 ⁇ M LacCer for 4 hours.
  • Mac-l was upregulated time-dependently by lOOnmol L LacCer (Figs. 10E) and maximum stimulation of Mac-1 level (2.5 fold compared to control) was observed 20 minutes after LacCer stimulation.
  • other GSL, GlcCer or ceramide did not alter Mac-1 expression on PMNs, indicating that this Mac-1 upregulation was a response specifically to LacCer (Figs. 10 K, L).
  • Figs. 10A-D show results of the following protocol.
  • PMNs were incubated with increasing concentrations of LacCer for 20 minutes, cooled rapidly on ice, washed and then incubated with either of monoclonal (m) anti-human (h) LFA-1, M-anti-h-MAC-1, m-anti-h pi 50,95 or m-anti-h L-selectin, followed by FITC- conjugated m-anti-mouse IgG.
  • the PMNs were gated by forward and side scattering and analyzed using FACscan.
  • E:PMNs were incubated with lOOnmol/L LacCer for indicated times and analyzed for Mac-1 expression.
  • Example 12- LacCer Stimulated PMNs Generate ROMs via NADPH Oxidase
  • LacCer activated superoxide generation via NADPH oxidase in human aortic smooth muscle cells, resulting in their proliferation.
  • ROM stimulation by LacCer in PMNs, using DCFH-DA was also evaluated.
  • a strikingly similar, positive, dose-dependent response was seen in these PMNs (Fig 11A).
  • This neutrophil ROM generation induced by 100 or 200nmol L LacCer (which had been sufficient to upregulate Mac-1 on PMNs), was inhibited by the antioxidant, NAC and completely blocked by the NADPH oxidase inhibitor, DPI (Fig 11 A).
  • DPI NADPH oxidase inhibitor
  • PMNs were first incubated in 96 well-plates with or without N- acetylcysteine (NAC) or diphenyleneiodonium (DPI) for 20 minutes at 37°C, then 2 , 7 - dichlorofluorescin diacetate and various concentrations of LacCer were added. This plate was incubated for 45 minutes at 37°C and read on a fluorescence plate reader. Data are means + SD for 6 wells. Each data point with inhibitor is significantly different from the point without inhibitor at the same concentration of LacCer (p ⁇ 0.001, *p ⁇ 0.01 by Student's t-test). LacCer enhanced ROM generation by PMN, and NAC or DPI inhibited this additional ROM generation.
  • NAC N- acetylcysteine
  • DPI diphenyleneiodonium
  • Fig. 1 IB PMNs were incubated with increasing concentrations of LacCer at 34°C, and lucigenin chemiluminescence was monitored for 20 minutes. Data are means + SD from three individual experiments. *p ⁇ 0.01 vs. no LacCer by ANOVA. LacCer enhanced this superoxide generation in a dose- and time-dependent manner. In Fig. 12C, LacCer induced superoxide generation was reduced by various inhibitors. In Fig. 12C PMNs were first incubated with several inhibitors of ROM for about 15 minutes.
  • Fig. 12C shows that LacCer alone (no inhibitor) abrogated superoxide levels about two-fold compared with controls. Pre-incubation with SOD completely abrogated LacCer induced superoxide generation. DPI and apocyanin were relatively less inhibitory with respect to superoxide generation in these PMNs.
  • PMNs were preincubated with each inhibitor for 15 minutes at room temperature and then stimulated with LacCer for 20 minutes. Superoxide generation was measured for 20 minutes in the absence (open bars) or presence (shaded bars) of lOOnmol/L LacCer. Data are means ⁇ SD of three individual experiments. *p ⁇ 0.01 vs. no inhibitor by Student's t-test.
  • SOD designates superoxide dismutase
  • DPI designates diphenyleneiodonium chloride.
  • the LacCer-induced lucigenin chemiluminescence was blocked by SOD, and inhibited by the NAPDH oxidase inhibitors.
  • PLA2 phospholipase A 2
  • Figs. 12A-B PMNs were first incubated with one of the inhibitors for 20 minutes, and then stimulated with lOOnmol/L LacCer for 20 minutes.
  • the inhibitors tested were DMSO only (vehicle), NAC, DPI, BATPA/AM, quinacrine, genistein, staurosporine and WEB 2086.
  • the Mac-1 expression was analyzed by FACScan.
  • Fig. 17 shows a model depicting LacCer-mediated redox signaling leading to ICAM-1 expression in endothelial cells and adhesion of neutrophils.
  • Figs. 18-19 summarize results of examples above and show LacCer-related pathways modulating cell proliferation (Figs. 18, 19) and cell adhesion (Fig. 19).
  • the invention has been described in detail with reference to preferred embodiments thereof. However, it will be appreciated that those skilled in the art, upon consideration of this disclosure, may make modification and improvements within the spirit and scope of the invention as set forth in the following claims.
  • ADDRESSEE Dike, Bronstein, Roberts & Cushman, LLP

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Urology & Nephrology (AREA)
  • Dermatology (AREA)
  • Vascular Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

La présente invention concerne des procédés de traitement et de prophylaxie de maladies, de troubles post-chirurgicaux et d'infections bactériennes associés au lactosylcéramide. Ces procédés consistent généralement à administrer à un mammifère, en particulier à un être humain, une quantité efficace, dans la perspective thérapeutique, d'un composé qui inhibe le UDPGal:GlcCerβ1-⊃4galactosylcéramide (GalT-2). L'invention concerne également des dosages biologiques in vitro et in vivo permettant de détecter des composés qui, du point de vue thérapeutique, sont susceptibles de moduler le GalT2.
PCT/US1998/009958 1997-05-21 1998-05-14 Procedes de traitement d'etats associes au lactosylceramide WO1998052553A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA2290742A CA2290742C (fr) 1997-05-21 1998-05-14 Utilisation d'un compose inhibiteur du galt-2 pour le traitement de maladies associees au lactosylceramide
AU76866/98A AU742468B2 (en) 1997-05-21 1998-05-14 Methods for treatment of conditions associated with lactosylceramide
JP55046098A JP2001526683A (ja) 1997-05-21 1998-05-14 ラクトシルセラミド関連症状の治療方法
EP98924776A EP0996433A4 (fr) 1997-05-21 1998-05-14 Procedes de traitement d'etats associes au lactosylceramide

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US4724997P 1997-05-21 1997-05-21
US08/998,262 US5972928A (en) 1997-05-21 1997-12-24 Methods for treatment of conditions associated with lactosylceramide
US60/047,249 1997-12-24
US08/998,262 1997-12-24

Publications (1)

Publication Number Publication Date
WO1998052553A1 true WO1998052553A1 (fr) 1998-11-26

Family

ID=26724804

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/009958 WO1998052553A1 (fr) 1997-05-21 1998-05-14 Procedes de traitement d'etats associes au lactosylceramide

Country Status (6)

Country Link
US (2) US5972928A (fr)
EP (1) EP0996433A4 (fr)
JP (1) JP2001526683A (fr)
AU (1) AU742468B2 (fr)
CA (1) CA2290742C (fr)
WO (1) WO1998052553A1 (fr)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999050385A2 (fr) * 1998-03-30 1999-10-07 President And Fellows Of Harvard College Regulation de la synthese de glycosaminoglycane, techniques apparentees et reactifs connexes
EP1098642A1 (fr) * 1998-07-27 2001-05-16 The Johns Hopkins University Methodes de traitement d'etats modules par la lactosylceramide
JP2002338469A (ja) * 2001-05-15 2002-11-27 Seikagaku Kogyo Co Ltd Vdac機能阻害剤
WO2009045503A1 (fr) * 2007-10-05 2009-04-09 Genzyme Corporation Procédé utilisant des dérivés de céramide pour traiter des maladies polykystiques des reins
US7615573B2 (en) 2001-07-16 2009-11-10 The Regents Of The University Of Michigan Synthesis of UDP-glucose: N-acylsphingosine glucosyltransferase inhibitors
WO2010039256A1 (fr) * 2008-10-03 2010-04-08 Genzyme Corporation Inhibiteurs de glucosylcéramide synthase de type 2-acylaminopropoanol
US8003617B2 (en) 2004-11-10 2011-08-23 Genzyme Corporation Methods of treating diabetes mellitus
US8304447B2 (en) 2007-05-31 2012-11-06 Genzyme Corporation 2-acylaminopropoanol-type glucosylceramide synthase inhibitors
US8389517B2 (en) 2008-07-28 2013-03-05 Genzyme Corporation Glucosylceramide synthase inhibition for the treatment of collapsing glomerulopathy and other glomerular disease
US8716327B2 (en) 2006-05-09 2014-05-06 Genzyme Corporation Methods of treating fatty liver disease
US10888547B2 (en) 2009-11-27 2021-01-12 Genzyme Corporation Amorphous and a crystalline form of genz 112638 hemitartrate as inhibitor of glucosylceramide synthase

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040204381A1 (en) * 1997-11-12 2004-10-14 Moskal Joseph R Detection and treatment of glyco-enzyme-related disease
US6713057B1 (en) * 1999-02-24 2004-03-30 The Johns Hopkins University Compositions and methods for modulating serum cholesterol
EP1384719A1 (fr) * 1999-07-09 2004-01-28 The Regents of The University of Michigan Inhibiteurs de l'enzyme synthetase de glucosyl-ceramide pour soigner le cancer, la sphingolipidose et les infections microbiennes
WO2002062777A2 (fr) * 2001-01-10 2002-08-15 The Regents Of The University Of Michigan Composes du type amino-ceramide et procedes therapeutiques d'utilisation
WO2003005965A2 (fr) * 2001-07-11 2003-01-23 Musc Foundation For Research Development Modulateurs de ceramidase et procedes d'utilisation associes
US6783541B2 (en) * 2001-11-21 2004-08-31 Miravant Medical Technologies, Inc. Methods for inhibiting or suppressing stenosis of arteriovenous access fistulas and grafts
US20060217560A1 (en) * 2002-04-29 2006-09-28 Shayman James A Amino ceramide-like compounds and therapeutic methods of use
US6916802B2 (en) * 2002-04-29 2005-07-12 Genzyme Corporation Amino ceramide-like compounds and therapeutic methods of use
WO2005123055A2 (fr) * 2004-06-14 2005-12-29 Musc Foundation For Research Development Methodes pour traiter des troubles inflammatoires
WO2011099980A1 (fr) 2010-02-12 2011-08-18 The Johns Hopkins University Utilisation de l'isoforme b1,4gait-v de la lactosylcéramide synthase, comme marqueur biologique du cancer
US20150118221A1 (en) * 2012-05-08 2015-04-30 The John Hopkins University Compositions and methods for treating cardiac hypertrophy
DE102014210662A1 (de) * 2014-06-04 2015-12-17 Gea Westfalia Separator Group Gmbh Vorrichtung und Verfahren zur Gewinnung von Glycoglycerolipiden und Glycosphingolipiden aus lipoiden Phasen

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5041441A (en) * 1988-04-04 1991-08-20 The Regents Of The University Of Michigan Method of chemotherapy using 1-phenyl-2-decanoylamino-3-morpholino-1-propanol

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5302609A (en) * 1992-12-16 1994-04-12 The Regents Of The University Of Michigan Treatment of diabetic nephropathy
US5707649A (en) * 1993-08-13 1998-01-13 Seikagaku Corporation Agent for treating neuronal diseases
WO1997010817A1 (fr) * 1995-09-20 1997-03-27 The Regents Of The University Of Michigan Composes de type amino ceramide et leurs methodes d'utilisation therapeutiques
NO965193L (no) * 1995-12-08 1997-06-09 Seikagaku Kogyo Kk Seikagaku C Aminalkoholderivat og fremgangsmåte for fremstilling derav
JP4140984B2 (ja) * 1995-12-20 2008-08-27 生化学工業株式会社 分化誘導作用を有する薬剤

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5041441A (en) * 1988-04-04 1991-08-20 The Regents Of The University Of Michigan Method of chemotherapy using 1-phenyl-2-decanoylamino-3-morpholino-1-propanol

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999050385A2 (fr) * 1998-03-30 1999-10-07 President And Fellows Of Harvard College Regulation de la synthese de glycosaminoglycane, techniques apparentees et reactifs connexes
WO1999050385A3 (fr) * 1998-03-30 1999-11-18 Harvard College Regulation de la synthese de glycosaminoglycane, techniques apparentees et reactifs connexes
EP1098642A4 (fr) * 1998-07-27 2004-08-18 Univ Johns Hopkins Methodes de traitement d'etats modules par la lactosylceramide
EP1098642A1 (fr) * 1998-07-27 2001-05-16 The Johns Hopkins University Methodes de traitement d'etats modules par la lactosylceramide
JP2002338469A (ja) * 2001-05-15 2002-11-27 Seikagaku Kogyo Co Ltd Vdac機能阻害剤
US7763738B2 (en) 2001-07-16 2010-07-27 Genzyme Corporation Synthesis of UDP-glucose: N-acylsphingosine glucosyltransferase inhibitors
US7615573B2 (en) 2001-07-16 2009-11-10 The Regents Of The University Of Michigan Synthesis of UDP-glucose: N-acylsphingosine glucosyltransferase inhibitors
US8779163B2 (en) 2001-07-16 2014-07-15 Genzyme Corporation Synthesis of UDP-Glucose: N-acylsphingosine glucosyl transferase inhibitors
US8138353B2 (en) 2001-07-16 2012-03-20 Genzyme Corporation Synthesis of UDP-glucose: N-acylsphingosine glucosyltransferase inhibitors
US9546161B2 (en) 2001-07-16 2017-01-17 Genzyme Corporation Synthesis of UDP-glucose: N-acylsphingosine glucosyl transferase inhibitors
US9532976B2 (en) 2004-11-10 2017-01-03 Genzyme Corporation Method of lowering blood glucose
US8003617B2 (en) 2004-11-10 2011-08-23 Genzyme Corporation Methods of treating diabetes mellitus
US8716327B2 (en) 2006-05-09 2014-05-06 Genzyme Corporation Methods of treating fatty liver disease
US9556155B2 (en) 2006-05-09 2017-01-31 Genzyme Corporation Methods of treating fatty liver disease
US8940776B2 (en) 2007-05-31 2015-01-27 Genzyme Corporation 2-acylaminopropoanol-type glucosylceramide synthase inhibitors
US9745294B2 (en) 2007-05-31 2017-08-29 Genzyme Corporation 2-acylaminopropoanol-type glucosylceramide synthase inhibitors
US8304447B2 (en) 2007-05-31 2012-11-06 Genzyme Corporation 2-acylaminopropoanol-type glucosylceramide synthase inhibitors
AU2008307516B2 (en) * 2007-10-05 2011-11-17 Genzyme Corporation Method of treating polycystic kidney diseases with ceramide derivatives
EP2550965A3 (fr) * 2007-10-05 2013-05-15 Genzyme Corporation Dérivés de céramide pour le traitement de la polykystose rénale
KR101640263B1 (ko) * 2007-10-05 2016-07-15 젠자임 코포레이션 세라마이드 유도체로 다낭성 신장질환을 치료하는 방법
KR20100084647A (ko) * 2007-10-05 2010-07-27 겐자임 코포레이션 세라마이드 유도체로 다낭성 신장질환을 치료하는 방법
US10220039B2 (en) 2007-10-05 2019-03-05 Genzyme Corporation Method of treating polycystic kidney diseases with ceramide derivatives
US8912177B2 (en) 2007-10-05 2014-12-16 Genzyme Corporation Method of treating polycystic kidney diseases with ceramide derivatives
WO2009045503A1 (fr) * 2007-10-05 2009-04-09 Genzyme Corporation Procédé utilisant des dérivés de céramide pour traiter des maladies polykystiques des reins
US8729075B2 (en) 2008-07-28 2014-05-20 Genzyme Corporation Glucosylceramide synthase inhibition for the treatment of collapsing glomerulopathy and other glomerular disease
US9481671B2 (en) 2008-07-28 2016-11-01 Genzyme Corporation Glucosylceramide synthase inhibition for the treatment of collapsing glomerulopathy and other glomerular disease
US8389517B2 (en) 2008-07-28 2013-03-05 Genzyme Corporation Glucosylceramide synthase inhibition for the treatment of collapsing glomerulopathy and other glomerular disease
US8309593B2 (en) 2008-10-03 2012-11-13 Genzyme Corporation 2-acylaminopropoanol-type glucosylceramide synthase inhibitors
EP3078373A1 (fr) * 2008-10-03 2016-10-12 Genzyme Corporation Inhibiteurs de glucosylcéramide synthase de type 2-acylaminopropanol
KR101687039B1 (ko) * 2008-10-03 2016-12-15 젠자임 코포레이션 2-아실아미노프로판올형 글루코실세라미드 합성효소 억제제
US9272996B2 (en) 2008-10-03 2016-03-01 Genzyme Corporation 2-acylaminopropoanol-type glucosylceramide synthase inhibitors
RU2578947C2 (ru) * 2008-10-03 2016-03-27 Джензайм Корпорейшн Ингибиторы глюкозилцерамидсинтазы 2-ациламинопропанольного типа
CN102271678A (zh) * 2008-10-03 2011-12-07 简詹姆公司 2-酰胺基丙醇型葡糖神经酰胺合成酶抑制剂
CN102271678B (zh) * 2008-10-03 2017-06-30 简詹姆公司 2‑酰胺基丙醇型葡糖神经酰胺合成酶抑制剂
US9744153B2 (en) 2008-10-03 2017-08-29 Genzyme Corporation 2-acylaminopropoanol-type glucosylceramide synthase inhibitors
KR20110067055A (ko) * 2008-10-03 2011-06-20 겐자임 코포레이션 2-아실아미노프로판올형 글루코실세라미드 합성효소 억제제
CN107935983A (zh) * 2008-10-03 2018-04-20 简詹姆公司 2‑酰胺基丙醇型葡糖神经酰胺合酶抑制剂
WO2010039256A1 (fr) * 2008-10-03 2010-04-08 Genzyme Corporation Inhibiteurs de glucosylcéramide synthase de type 2-acylaminopropoanol
US10888547B2 (en) 2009-11-27 2021-01-12 Genzyme Corporation Amorphous and a crystalline form of genz 112638 hemitartrate as inhibitor of glucosylceramide synthase
US11458119B2 (en) 2009-11-27 2022-10-04 Genzyme Corporation Amorphous and a crystalline form of genz 112638 hemitartrate as inhibitor of glucosylceramide synthase

Also Published As

Publication number Publication date
CA2290742A1 (fr) 1998-11-26
EP0996433A4 (fr) 2009-05-27
AU742468B2 (en) 2002-01-03
EP0996433A1 (fr) 2000-05-03
CA2290742C (fr) 2010-08-17
US5972928A (en) 1999-10-26
JP2001526683A (ja) 2001-12-18
AU7686698A (en) 1998-12-11
US6228889B1 (en) 2001-05-08

Similar Documents

Publication Publication Date Title
AU742468B2 (en) Methods for treatment of conditions associated with lactosylceramide
US6511979B1 (en) Methods for treating conditions modulated by lactosylceramide
Qiu et al. Hyperammonemia-mediated autophagy in skeletal muscle contributes to sarcopenia of cirrhosis
Booth et al. Elevated ambient glucose induces acute inflammatory events in the microvasculature: effects of insulin
CA2147881C (fr) Dithiocarbamates pour le traitement de l'atherosclerose et autres maladies cardiovasculaires et inflammatoires
Bruzzese et al. Synergistic antitumor effect between vorinostat and topotecan in small cell lung cancer cells is mediated by generation of reactive oxygen species and DNA damage-induced apoptosis
US20150044193A1 (en) COMPOSITIONS AND METHODS FOR THE TREATMENT OF ALTERED a-SYNUCLEIN FUNCTION
Modrak et al. Ceramide regulates gemcitabine-induced senescence and apoptosis in human pancreatic cancer cell lines
US8785383B2 (en) Mast cell stabilizers in the treatment of obesity
US5792787A (en) Treatment for atherosclerosis and other cardiovascular and inflammatory diseases
Kamanna et al. Oxidized low-density lipoprotein stimulates monocyte adhesion to glomerular endothelial cells
CA2755191A1 (fr) Inhibiteurs de la liaison a une proteine kinase
Kim et al. Syringaresinol reverses age-related skin atrophy by suppressing FoxO3a-mediated matrix metalloproteinase–2 activation in Copper/Zinc superoxide dismutase–deficient mice
Pignatelli et al. Vitamin C inhibits platelet expression of CD40 ligand
KR20010101790A (ko) (e)-7-[4-(4-플루오로페닐)-6-이소프로필-2-[메틸(메틸술포닐)아미노]피리미딘-5-일](3r,5s)-3,5-디히드록시헵트-6-엔산 및 p450 이소효소 3a4의 억제제, 유도제 또는기질을 함유하는 약물 조합물
Lodhi et al. Evaluation of mechanism of hepatotoxicity of leflunomide using albino wistar rats
Yang et al. Sphingosine-1-phosphate induces myocyte autophagy after myocardial infarction through mTOR inhibition
Song et al. l‐Asparaginase‐mediated downregulation of c‐Myc promotes 1, 25 (OH) 2D3‐induced myeloid differentiation in acute myeloid leukemia cells
Huang et al. Molecular mechanisms of lymphocyte extravasation. III. The loss of lymphocyte extravasation potential induced by pertussis toxin is not mediated via the activation of protein kinase C.
Cekic et al. The effect of vitamin C on amiodarone-induced toxicity in rat thymocytes
Emanueli et al. Characterization of the endothelin receptor subtype mediating epithelium‐derived relaxant nitric oxide release from guinea‐pig trachea
EP4132500A1 (fr) Traitement combinatoire pour le lymphome
EP1825853A2 (fr) Procédés pour le traitement des affections modulées par le lactosylcéramide
Nakaue et al. Mechanisms of preferential bone formation in myeloma bone lesions by proteasome inhibitors
Hamasaki et al. (9-[4-acetyl-3-hydroxy-2-n-propylphenoxy) methyl]-3-(1H-tetrazol-5-yl)-4H-pyrido [1, 2-a] pyrimidin-4-one), AS-35, inhibits leukotriene synthesis

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2290742

Country of ref document: CA

Kind code of ref document: A

Ref document number: 1998 550460

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1998924776

Country of ref document: EP

Ref document number: 76866/98

Country of ref document: AU

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1998924776

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 76866/98

Country of ref document: AU